Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Brief Communication
  • Published:

Overexpression of Hmga2 activates Igf2bp2 and remodels transcriptional program of Tet2-deficient stem cells in myeloid transformation

Abstract

High Mobility Group AT-hook 2 (HMGA2) is a chromatin modifier and its overexpression has been found in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Level of Hmga2 expression is fine-tuned by Lin28b-Let-7 axis and Polycomb Repressive Complex 2, in which deletion of Ezh2 leads to activation of Hmga2 expression in hematopoietic stem cells. To elucidate the mechanisms by which the overexpression of HMGA2 helps transformation of stem cells harboring a driver mutation of TET2, we generated an Hmga2-expressing Tet2-deficient mouse model showing the progressive phenotypes of MDS and AML. The overexpression of Hmga2 remodeled the transcriptional program of Tet2-deficient stem and progenitor cells, leading to the impaired differentiation of myeloid cells. Furthermore, Hmga2 was bound to a proximal region of Igf2bp2 oncogene, and activated its transcription, leading to enhancing self-renewal of Tet2-deficient stem cells that was suppressed by inhibition of the DNA binding of Hmga2. These combinatory effects on the transcriptional program and cellular function were not redundant to those in Tet2-deficient cells. The present results elucidate that Hmga2 targets key oncogenic pathways during the transformation and highlight the Hmga2-Igf2bp2 axis as a potential target for therapeutic intervention.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Hmga2 overexpression and Tet2 loss induce the development of MDS and AML in vivo.
Fig. 2: Hmga2-expressing Tet2-deficient HSPCs enhance the repopulating capacity and impair the myeloid differentiation.
Fig. 3: Hmga2 overexpression and Tet2 loss result in the accumulation of alterations in gene expression.
Fig. 4: Hmga2 activates expression of Igf2bp and promotes self-renewal of Tet2-deficient HSPCs.

References

  1. Nimer SD. Myelodysplastic syndromes. Blood. 2008;111:4841–51.

    Article  CAS  Google Scholar 

  2. Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12:599–612.

    Article  CAS  Google Scholar 

  3. Sperling AS, Gibson CJ, Ebert BL. The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia. Nat Rev Cancer. 2017;17:5–19.

    Article  CAS  Google Scholar 

  4. Moran-Crusio K, Reavie L, Shih A, Abdel-Wahab O, Ndiaye-Lobry D, Lobry C, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011;20:11–24.

    Article  CAS  Google Scholar 

  5. Quivoron C, Couronné L, Della Valle V, Lopez CK, Plo I, Wagner-Ballon O, et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell. 2011;20:25–38.

    Article  CAS  Google Scholar 

  6. Muto T, Sashida G, Oshima M, Wendt GR, Mochizuki-Kashio M, Nagata Y, et al. Concurrent loss of Ezh2 and Tet2 cooperates in the pathogenesis of myelodysplastic disorders. J Exp Med. 2013;210:2627–39.

    Article  CAS  Google Scholar 

  7. Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210:2641–59.

    Article  CAS  Google Scholar 

  8. Hasegawa N, Oshima M, Sashida G, Matsui H, Koide S, Saraya A, et al. Impact of combinatorial dysfunctions of Tet2 and Ezh2 on the epigenome in the pathogenesis of myelodysplastic syndrome. Leukemia. 2017;31:861–71.

    Article  CAS  Google Scholar 

  9. Thanos D, Maniatis T. The High Mobility Group protein HMG I(Y) is required for NF-κB-dependent virus induction of the human IFN-β gene. Cell. 1992;71:777–89.

    Article  CAS  Google Scholar 

  10. Chin MT, Pellacani A, Wang H, Lin SSJ, Jain MK, Perrella MA, et al. Enhancement of serum-response factor-dependent transcription and DNA binding by the architectural transcription factor HMG-I(Y). J Biol Chem. 1998;273:9755–60.

    Article  CAS  Google Scholar 

  11. Fusco A, Fedele M. Roles of HMGA proteins in cancer. Nat Rev Cancer. 2007;7:899–910.

    Article  CAS  Google Scholar 

  12. Nishino J, Kim I, Chada K, Morrison SJ. Hmga2 promotes neural stem cell self-renewal in Young but not old mice by reducing p16Ink4a and p19Arf expression. Cell. 2008;135:227–39.

    Article  CAS  Google Scholar 

  13. Kishi Y, Fujii Y, Hirabayashi Y, Gotoh Y. HMGA regulates the global chromatin state and neurogenic potential in neocortical precursor cells. Nat Neurosci. 2012;15:1127–33.

    Article  CAS  Google Scholar 

  14. Copley MR, Babovic S, Benz C, Knapp DJHF, Beer PA, Kent DG, et al. The Lin28b-let-7-Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells. Nat Cell Biol. 2013;15:916–25.

    Article  CAS  Google Scholar 

  15. Oguro H, Yuan J, Tanaka S, Miyagi S, Mochizuki-Kashio M, Ichikawa H, et al. Lethal myelofibrosis induced by Bmi1-deficient hematopoietic cells unveils a tumor suppressor function of the polycomb group genes. J Exp Med. 2012;209:445–54.

    Article  CAS  Google Scholar 

  16. Rowe RG, Wang LD, Coma S, Han A, Mathieu R, Pearson DS, et al. Developmental regulation of myeloerythroid progenitor function by the Lin28b-let-7-Hmga2 axis. J Exp Med. 2016;213:1497–512.

    Article  CAS  Google Scholar 

  17. Cabezas-Wallscheid N, Klimmeck D, Hansson J, Lipka DB, Reyes A, Wang Q, et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell. 2014;15:507–22.

    Article  CAS  Google Scholar 

  18. Oshima M, Hasegawa N, Mochizuki-Kashio M, Muto T, Miyagi S, Koide S, et al. Ezh2 regulates the Lin28/let-7 pathway to restrict activation of fetal gene signature in adult hematopoietic stem cells. Exp Hematol. 2016;44:282–96.

    Article  CAS  Google Scholar 

  19. Sashida G, Wang C, Tomioka T, Oshima M, Aoyama K, Kanai A, et al. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J Exp Med. 2016;213:1459–77.

    Article  CAS  Google Scholar 

  20. Shimizu T, Kubovcakova L, Nienhold R, Zmajkovic J, Meyer SC, Shen HH, et al. Loss of Ezh2 synergizes with JAK2 -V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis. J Exp Med. 2016;213:1479–96.

    Article  CAS  Google Scholar 

  21. Dutta A, Hutchison RE, Mohi G. Hmga2 promotes the development of myelofibrosis in Jak2V617F knockin mice by enhancing TGF-beta1 and Cxcl12 pathways. Blood. 2017;130:920–32.

    Article  CAS  Google Scholar 

  22. Guglielmelli P, Zini R, Bogani C, Salati S, Pancrazzi A, Bianchi E, et al. Molecular profiling of CD34+ cells in idiopathic myelofibrosis identifies a set of disease-associated genes and reveals the clinical significance of Wilms’ tumor gene 1 (WT1). Stem Cells. 2007;25:165–73.

    Article  CAS  Google Scholar 

  23. Marquis M, Beaubois C, Lavallée VP, Abrahamowicz M, Danieli C, Lemieux S, et al. High expression of HMGA2 independently predicts poor clinical outcomes in acute myeloid leukemia. Blood Cancer J. 2018;8.68.

  24. Ory DS, Neugeboren BA, Mulligan RC. A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes. Proc Natl Acad Sci USA. 1996;93:11400–6.

    Article  CAS  Google Scholar 

  25. Sternberg A, Killick S, Littlewood T, Hatton C, Peniket A, Seidl T, et al. Evidence for reduced B-cell progenitors in early (low-risk) myelodysplastic syndrome. Blood. 2005;106:2982–91.

    Article  CAS  Google Scholar 

  26. Pellagatti A, Cazzola M, Giagounidis A, Perry J, Malcovati L, Della Porta MG, et al. Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells. Leukemia. 2010;24:756–64.

    Article  CAS  Google Scholar 

  27. Pellagatti A, Armstrong RN, Steeples V, Sharma E, Repapi E, Singh S, et al. Impact of spliceosome mutations on RNA splicing in myelodysplasia: dysregulated genes/pathways and clinical associations. Blood. 2018;132:1225–40.

    Article  CAS  Google Scholar 

  28. Zhou T, Kinney MC, Scott LM, Zinkel SS, Rebel VI. Revisiting the case for genetically engineered mouse models in human myelodysplastic syndrome research. Blood. 2015;126:1057–68.

    Article  CAS  Google Scholar 

  29. Yokomizo-Nakano T, Kubota S, Bai J, Hamashima A, Morii M, Sun Y, et al. Overexpression of RUNX3 represses RUNX1 to drive transformation of myelodysplastic syndrome. Cancer Res. 2020;80:2523–36.

    Article  CAS  Google Scholar 

  30. Degrauwe N, Schlumpf TB, Janiszewska M, Martin P, Cauderay A, Provero P, et al. The RNA binding protein IMP2 preserves glioblastoma stem cells by preventing let-7 target gene silencing. Cell Rep. 2016;15:1634–47.

    Article  CAS  Google Scholar 

  31. Dai N, Ji F, Wright J, Minichiello L, Sadreyev R, Avruch J. IGF2 mRNA binding protein-2 is a tumor promoter that drives cancer proliferation through its client mRNAs IGF2 and HMGA1. Elife. 2017;6:e27155.

  32. Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20:285–95.

    Article  CAS  Google Scholar 

  33. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74.

    Article  Google Scholar 

  34. Watanabe-Okochi N, Kitaura J, Ono R, Harada H, Harada Y, Komeno Y, et al. AML1 mutations induced MDS and MDS/AML in a mouse BMT model. Blood. 2008;111:4297–308.

    Article  CAS  Google Scholar 

  35. Cleynen I, Brants JR, Peeters K, Deckers R, Debiec-Rychter M, Sciot R, et al. HMGA2 regulates transcription of the Imp2 gene via an intronic regulatory element in cooperation with nuclear factor-κB. Mol Cancer Res. 2007;5:363–72.

    Article  CAS  Google Scholar 

  36. He X, Li W, Liang X, Zhu X, Zhang L, Huang Y, et al. IGF2BP2 overexpression indicates poor survival in patients with acute myelocytic leukemia. Cell Physiol Biochem. 2018;51:1945–56.

    Article  CAS  Google Scholar 

  37. Shastri A, Will B, Steidl U, Verma A. Stem and progenitor cell alterations in myelodysplastic syndromes. Blood. 2017;129:1586–94.

    Article  CAS  Google Scholar 

  38. Platzbecker U, Germing U, Götze KS, Kiewe P, Mayer K, Chromik J, et al. Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol. 2017;18:1338–47.

    Article  CAS  Google Scholar 

  39. Zhang X, Su J, Jeong M, Ko M, Huang Y, Park HJ, et al. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat Genet. 2016;48:1014–23.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank the members of the Sashida Laboratory for their discussions during the preparation of this manuscript and Mr. Shinji Kudoh and Dr. Takaaki Ito for their technical help. This work was supported in part by a grant from the Daiichi Sankyo Foundation of Life Science (to GS), the Uehara Memorial Foundation (to GS), the Princess Takamatsu Cancer Research Fund (to GS), the Japanese Society of Hematology (to GS), International Joint Usage/Research Center, the Institute of Medical Science, the University of Tokyo (to GS), and Grants-in-Aid for Scientific Research (16KT0113, 18H02842, 19K22640 and 19K08842) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Goro Sashida.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bai, J., Yokomizo-Nakano, T., Kubota, S. et al. Overexpression of Hmga2 activates Igf2bp2 and remodels transcriptional program of Tet2-deficient stem cells in myeloid transformation. Oncogene 40, 1531–1541 (2021). https://doi.org/10.1038/s41388-020-01629-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-020-01629-w

This article is cited by

Search

Quick links